WO2016191395A1 - Treatment of post-bariatric hypoglycemia with exendin(9-39) - Google Patents

Treatment of post-bariatric hypoglycemia with exendin(9-39) Download PDF

Info

Publication number
WO2016191395A1
WO2016191395A1 PCT/US2016/033837 US2016033837W WO2016191395A1 WO 2016191395 A1 WO2016191395 A1 WO 2016191395A1 US 2016033837 W US2016033837 W US 2016033837W WO 2016191395 A1 WO2016191395 A1 WO 2016191395A1
Authority
WO
WIPO (PCT)
Prior art keywords
exendin
morning
injectate
administration
concentration
Prior art date
Application number
PCT/US2016/033837
Other languages
English (en)
French (fr)
Inventor
Tracey L. MCLAUGHLIN
Colleen M. CRAIG
Original Assignee
The Bot Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to PL16800621T priority Critical patent/PL3297654T3/pl
Priority to US15/576,647 priority patent/US10639354B2/en
Priority to ES16800621T priority patent/ES2887723T3/es
Priority to BR112017025000-4A priority patent/BR112017025000A2/pt
Priority to LTEPPCT/US2016/033837T priority patent/LT3297654T/lt
Priority to EP21176474.1A priority patent/EP3936143A1/en
Priority to CA3024358A priority patent/CA3024358A1/en
Application filed by The Bot Of The Leland Stanford Junior University filed Critical The Bot Of The Leland Stanford Junior University
Priority to SI201631300T priority patent/SI3297654T1/sl
Priority to EP16800621.1A priority patent/EP3297654B1/en
Priority to HRP20211285TT priority patent/HRP20211285T1/hr
Priority to RS20211073A priority patent/RS62368B1/sr
Priority to DK16800621.1T priority patent/DK3297654T3/da
Priority to AU2016267057A priority patent/AU2016267057B2/en
Publication of WO2016191395A1 publication Critical patent/WO2016191395A1/en
Priority to US16/813,535 priority patent/US10993991B2/en
Priority to US17/226,164 priority patent/US11617782B2/en
Priority to CY20211100779T priority patent/CY1124616T1/el
Priority to AU2022201973A priority patent/AU2022201973A1/en
Priority to US18/172,546 priority patent/US20240000894A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2278Vasoactive intestinal peptide [VIP]; Related peptides (e.g. Exendin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis

Definitions

  • the present invention provides methods and compositions for the treatment of hypoglycemia, particularly post-bariatric hyperinsulinemia, and more generally hyperinsulinemic hypoglycemia of any origin, and the prevention of associated acute symptoms and chronic outcomes in which a glucagon-like peptide-1 receptor antagonist (GLP1A), exendin(9-39), is subcutaneously administered in a therapeutically effective dose.
  • GLP1A glucagon-like peptide-1 receptor antagonist
  • exendin(9-39) exendin(9-39
  • GLP-1 glucagon-like peptide-1
  • the present invention relates to pharmaceutical compositions and methods involving twice-per-day subcutaneous delivery of exendin(9-39) in doses therapeutically effective for treating or preventing hyperinsulinemic hypoglycemia in a patient who has previously had bariatric surgery, such as Roux-en-Y gastric bypass surgery.
  • the twice-per- day administration is generally a first administration in the morning and a second administration in the evening.
  • the twice-per-day subcutaneous delivery involves administering a composition comprising a therapeutically effective amount of exendin(9-39), wherein the therapeutically effective amount is in the range of 10 mg-30 mg exendin(9-39).
  • the therapeutically effective amount of exendin(9- 39) administered twice-per-day is in the range of 10-20 mg (e.g., 10 mg, 15 mg, or 20 mg), in the range of 10 mg-15 mg (e.g., 10 mg or 15 mg), or in the range of 15 mg– 30 mg (e.g., 15 mg, 20 mg, 25 mg, or 30 mg).
  • the exendin(9-39)-containing composition may be a solution or suspension that comprises exendin(9-39) at a concentration in the range of 4-20 mg/ml. In some embodiments the concentration is in the range of 10-20 mg/ml. In some embodiments the concentration is in the range of 8-16 mg/ml. In some embodiments the concentration is in the range of 13-16 mg/ml.
  • the composition, or injectate, administered in the morning administration and the composition, or injectate, administered in the evening administration are the same, i.e., they contain the same amount (dose) of exendin(9-39) at the same concentration.
  • subcutaneous administration twice-per-day comprises administering a morning injectate and an evening injectate, where the amount and/or concentration of exendin(9-39) in the evening injectate is greater than the amount of exendin(9-39) in the morning injectate.
  • the amount of exendin(9-39) in the evening injectate is greater than the amount of exendin(9-39) in the morning injectate.
  • the concentration of exendin(9-39) in the evening injectate is higher than the concentration of exendin(9-39) in the morning injectate.
  • the amount of exendin(9-39) in the evening injectate is 5 mg-10 mg greater than the amount in the morning injectate. In some such cases the amount of exendin(9-39) in the morning injectate is 10 mg and the amount in the evening injectate is 15 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 10 mg and the amount in the evening injectate is 20 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 15 mg and the amount in the evening injectate is 20 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 15 mg and the amount in the evening injectate is 25 mg.
  • the amount of exendin(9-39) in the morning injectate is 20 mg and the amount in the evening injectate is 25 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 20 mg and the amount in the evening injectate is 30 mg.
  • the concentration of exendin(9-39) in the morning injectate is not the same as the concentration of exendin(9-39) in the evening injectate.
  • the concentration of exendin(9-39) in morning injectate is 15 mg/ml and the concentration of exendin(9-39) in the evening injectate is 20 mg/ml.
  • the exendin(9-39) amount and concentration of the morning and evening injectates are selected such that the exendin(9-39) Tmax after the evening administration is longer than the Tmax after the morning administration.
  • the amount and concentration of the morning and evening injectates are selected such that the administration results in an exendin(9-39) Cmax of at least 100 ng/ml (e.g., as measured by liquid chromatograph-mass spectrometry).
  • administration comprises a first daily administration is in the morning and a second daily administration in the evening, where the second daily administration is at least 8 hours after the first daily administration.
  • the morning administration is administered before the morning meal (e.g., breakfast).
  • the morning administration is administered at least 1 hour before the morning meal, optionally 60 to 90 minutes before the morning meal.
  • the morning administration is in the period from one hour before the morning meal to one hour after the morning meal.
  • the second daily administration is administered 60 to 90 minutes before the evening meal.
  • the second daily administration is administered after the evening meal and before bedtime (for example, within 2 hours of bedtime).
  • the evening administration is from 9 to 15 hours after the morning administration.
  • a dose of exendin(9-39) is administered with regard to the timing of a meal.
  • the dose of exendin(9-39) is administered with a meal, or before a particular meal, including, for example a certain time, e.g., 15 minutes to two hours, e.g. one hour, before a meal, or a certain time after a meal.
  • a dose of exendin(9-39) is administered without regard to the timing of a meal.
  • the patient self-administers (or is wearing a device programmed to administer) the exendin(9-39).
  • the first dose may be administered in the evening, such that it provides protection during breakfast the following day, with subsequent doses following the next morning and the next evening about twelve hours later.
  • exendin(9-39) is formulated and administered in an injectable pen device or via a vial/syringe combination that may be pre-programmed or marked to deliver a fixed dosage amount (and optionally two different fixed dosage amounts corresponding to morning and evening administrations) ranging from 10-30 mg exendin(9- 39).
  • compositions of the invention have use for treatment and prevention of hyperinsulinemic hypoglycemia and its associated symptoms and outcomes in patients with hyperinsulinemic hypoglycemia post bariatric surgery and post gastrointestinal surgery.
  • the invention provides for the prevention and treatment of associated acute and chronic symptoms and outcomes in susceptible patients. Treatment in accordance with the invention of patients in need of therapy will improve patient quality of life both in the short- and long-term, will reduce overall patient morbidity, and may prevent premature death and/or extend life-expectancy.
  • FIG. 1A-B Average plasma glucose (A) and insulin (B) responses to a 75 gram oral glucose tolerance test (OGTT) in subjects with hyperinsulinemic hypoglycemia during a randomized blinded cross-over study in which a primed continuous intravenous (IV) infusion of exendin(9-39) (at 500 pmol/kg/min over 180 minutes) or placebo (normal saline) was administered, as described in Example 1.
  • Solid line placebo infusion
  • dashed line exendin(9-39) infusion.
  • FIG. 2A-C Average plasma GLP-1 (A), GIP (B), and glucagon (C) response to a 75 gram OGTT in subjects with hyperinsulinemic hypoglycemia receiving primed continuous IV infusion of exendin(9-39) of 500 pmol/kg/min over 180 minutes versus placebo (normal saline) infusion, as described in Example 1. Solid line: placebo infusion; dashed line: exendin(9-39) infusion.
  • FIG.3 Individual and average symptomatic responses to a 75 gram OGTT in 8 patients with hyperinsulinemic hypoglycemia receiving a primed continuous IV infusion of exendin(9-39) versus placebo, as described in Example 1. Overall Symptom Score, Glucose Rise, and Glucose Fall Scores are presented. Continuous IV infusion of exendin(9-39) at 500 pmol/kg/min over 180 minutes substantially improved symptoms of hypoglycemia, as demonstrated by the reduced Overall Symptom and Glucose Fall scores.
  • FIG. 5A-B Average plasma exendin(9-39) concentrations for 8 human subjects administered a continuous exendin(9-39) IV infusion at a rate of 500 pmol/kg/min over 180 minutes are plotted (black line). The projected exendin(9-39) pharmacokinetic response to a single IV bolus of 7,500 pmol/kg exendin(9-39) administered at T-30 (blue line) was extrapolated based on the known half-life of intravenously administered exendin(9- 39).
  • FIG. 6 Average plasma glucose levels during a 75 gram OGTT for subjects administered a subcutaneous dose of exendin(9-39) as compared to baseline.
  • FIG. 7A-B Plasma exendin(9-39) concentrations following subcutaneous injection of exendin(9-39).
  • A Three subjects received a single subcutaneous injection of approximately 10, 20, or 30 mg of exendin(9-39) in a volume of 0.7 ml (5x, 10x, or 15x doses, respectively).
  • B Five subjects received doses of approximately 2, 10, 20, or 30 mg (1x, 5x, 10, or 15x, respectively), with each dose administered at a concentration of 15 mg/ml or less; higher doses were administered via more than one injection so as to maintain a relatively dilute concentration.
  • FIG. 8 Percent increase in plasma glucose nadir concentrations were calculated for the subcutaneously administered doses of exendin(9-39) relative to baseline. A correlation was observed between higher percent increases in plasma glucose nadir concentrations and increasing peak plasma exendin(9-39) concentrations (C max ).
  • FIG. 9 Study design for 3-day Multi-Ascending Dose Trial to assess the safety, tolerability, efficacy, and pharmacokinetic profile of BID exendin(9-39) administered subcutaneously over 3 days to patients with post-bariatric hyperinsulinemic hypoglycemia.
  • FIG. 10 Exendin(9-39) plasma concentrations on Day 3 after 5 doses as described in Example 4.
  • Table 1 Metabolic responses to a 75 gram oral glucose tolerance test (OGTT) during primed continuous IV infusion of exendin(9-39) in eight post-RYGB patients with hyperinsulinemic hypoglycemia (HH). Metabolic responses of eight BMI, age, and sex matched non-surgical controls are presented for comparison. AUC values were calculated by the trapezoidal rule utilizing the last value carried forward to account for prematurely discontinued OGTTs in cases of hypoglycemia, which occurred solely during placebo infusion.
  • OGTT oral glucose tolerance test
  • HH hyperinsulinemic hypoglycemia
  • Table 2 Mean plasma GLP-1, GIP and glucagon response to a 75 gram oral glucose tolerance test (OGTT) in eight patients with hyperinsulinemic hypoglycemia (HH) during a primed continuous IV infusion of exendin(9-39) of 500 pmol/kg/min over 180 minutes vs. during placebo (normal saline) infusion.
  • OGTT oral glucose tolerance test
  • Table 3 Subject metabolic and symptomatic response to a single subcutaneous (SC) injection of 10-30 mg of exendin(9-39), denoted as SC Ex(9-39), continuous IV infusion of exendin(9-39 (IV Ex(9-39)), or placebo during a 75 gram OGTT.
  • SC subcutaneous
  • IV Ex(9-39) continuous IV infusion of exendin(9-39)
  • placebo placebo during a 75 gram OGTT.
  • Table 4 PK/PD response to increasing doses/increasing concentrations.
  • subjects 2-5 each received a subcutaneous injection of exendin(9-39) in doses ranging from 37,500-112,500 pmol/kg (approximately 10-30 mg) each in a volume of 0.7ml, resulting in dose concentrations of approximately 15-40 mg/ml.
  • Shown here are subject PK/PD responses to each dose. Injectate concentrations of approximately 15 mg/ml resulted in the greatest pharmacodynamic response, as defined by nadir postprandial glucose and AUC glucose, and greatest pharmacokinetic response, as defined by Cmax and DN Cmax.
  • a relatively dilute dose may be preferred for BID dosing, and a more concentrated formulation may be preferred for less frequent dosing or a more sustained exposure.
  • the 75,000 pmol/kg dose (17 mg) with a concentration of about 24 mg/ml resulted in a favorable sustained/slow release pharmacokinetic response, with a half-life of 9.14 hours, and a Cmax that was 70 or more ng/ml.
  • a relatively concentrated dose may be used advantageously for qD or BID dosing not tied to meals.
  • Table 5 PK/PD response to increasing dose with constant injectate concentration. As described in Example 3, four subjects received subcutaneous injections of 37,500-112,500 pmol/kg exendin(9-39) in equivalent concentrations (approximately 13-16 mg/ml), as this concentration was found to result in a favorable immediate release formulation of the invention. Results shown demonstrate an increasingly favorable PK response with increasing dose, as defined by Cmax and T 1/2 .
  • Table 6 PK/PD response in four subjects dosed with varying doses of subcutaneously administered BID exendin(9-39) in a 3-day clinical trial as described in Example 4. DETAILED DESCRIPTION OF THE INVENTION
  • Post-bariatric hyperinsulinemic hypoglycemia is a disorder that is characterized by low blood sugar and elevated insulin levels 1-3 hours after meals.
  • the disorder manifests as neuroglycopenic symptoms (such as confusion, loss of focus, fatigue, ataxia, paralysis, seizures, or loss of consciousness), vasomotor symptoms (such as sweating and shakiness), and/or adrengeric symptoms (such as heart palpitations).
  • Exendin(9-39) administered intravenously is characterized by a short plasma half-life of 33 minutes (see, Gardiner et al., JPET 316:852-859 (2006); see also, Edwards et al., Diabetes 48:86-93 (1999)).
  • exendin(9-39) administered as a single IV bolus of 7,500 pmol/kg prevented hypoglycemia in patients only if the bolus was timed to closely coincide with peak GLP-1 plasma concentrations.
  • exendin(9-39) The pharmacokinetic properties exhibited from the administration of a single intravenous dose of exendin(9-39) are such that a person of ordinary skill in the art would not have expected subcutaneous administration of clinically appropriate doses of exendin(9-39) to be therapeutically effective in the treatment of postprandial hyperinsulinemic hypoglycemia.
  • exendin(9-39) by twice-per-day (BID) subcutaneous injection (SC) can effectively prevent hypoglycemia in patients having post-bariatric hyperinsulinemic hypoglycemia, that such prevention results at a dose of 30 mg or lower (e.g., in the range 7.5 mg-20 mg, e.g., 10 mg- 15 mg), that such prevention can be achieved using a convenient administration schedule not necessarily tied to meal times, and that the pharmacokinetics of exendin(9-39) SC administration may be tuned based on the concentration of exendin(9-39) in the injectate, as well as by dose, to achieve an efficacious treatment. See Example 3 and Example 4.
  • the present invention relates to pharmaceutical compositions and methods for subcutaneously administering exendin(9-39) at a BID dose in the range of about 7.5 mg– 20 mg for the treatment and prevention of hyperinsulinemic hypoglycemia.
  • the present invention represents a significant advance in the field of surgical intervention for weight loss and/or metabolic control. This is especially important, because those post-bariatric patients currently suffering hypoglycemic excursions have no effective therapy and are sometimes critically ill. The intractable nature of the problem has been highlighted by those patients with disease so debilitating they reversed the surgery, or underwent other highly morbid procedures, such as partial pancreatectomy, only to learn the condition persists.
  • the present invention provides a therapeutic intervention that can largely protect them should they suffer from post-bariatric hyperinsulinemia.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of shall mean excluding other elements that would materially affect the basic and novel characteristics of the claimed invention.
  • Consisting of shall mean excluding any element, step, or ingredient not specified in the claim. Embodiments defined by each of these transition terms are within the scope of this invention.
  • Exendin(9-39) or“Ex(9-39)” or“Ex9” refers to a 31 amino acid peptide with an empirical formula of C 149 H 234 N 40 O 47 S and a molecular weight of 3369.8 Daltons.
  • the amino acid sequence for exendin(9-39) is shown as follows: H-Asp-Leu-Ser-Lys-Gln-Met- Glu-Glu-Glu-Ala-Val-Arg-Leu-Phe-Ile-Glu-Trp-Leu-Lys-Asn-Gly-Gly-Pro-Ser-Ser-Gly- Ala-Pro-Pro-Pro-Ser-NH 2 .
  • Exendin(9-39) comprises residues 9-39 of the GLP-1 receptor agonist exendin-4 and is a GLP-1 receptor antagonist. See, Montrose-Rafizadeh et al., Journal of Biological Chemistry, 272:21201-21206 (1997).
  • exendin(9-39) encompasses pharmaceutically acceptable salts of exendin(9-39), including but not limited to sulfate, hydrochloride, phosophate, sulfamate, acetate, citrate, lactate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, cyclohexylsulfamate and quinate salts.
  • exendin(9-39) is in the form of exendin(9-39) acetate or exendin(9-39) trifluoroacetate. Where not otherwise specified herein, exendin(9-39) acetate is used (obtained from Bachem (Clinalfa, Läufelfingen, Switzerland)).
  • GLP1A refers to a GLP1 receptor antagonist (sometimes referred to as a“GLP1 antagonist”).
  • the terms“administer,”“administering,” and“administration,” as used herein, refer to introducing a compound (e.g., exendin(9-39)) or composition into a human subject.
  • the terms encompass both direct administration (e.g., administration to a subject by a medical professional or other caregiver, or by self-administration, or by programming an automatic device to deliver exendin(9-39) on a BID schedule) and indirect administration (e.g., the act of prescribing a compound or composition to a subject).
  • the terms“treatment,”“treating,” and“treat,” as used herein in reference to administering exendin(9-39) to treat hyperinsulinemic hypoglycemia covers any treatment of a disease in a human subject, and includes: (a) reducing the risk, frequency or severity of hypoglycemic episodes in patients with a history of hyperinsulinemic hypoglycemia, (b) reducing the risk of occurrence of hypoglycemia in a subject determined to be predisposed to the disease, such as a person who has received post-bariatric surgery, but not yet diagnosed as having the disease, (c) impeding the development of the disease; and/or (d) relieving the disease, i.e., causing regression of the disease and/or relieving one or more disease symptoms.
  • injectate refers the exendin(9-39)-containing composition subcutaneously delivered to a patient at a morning or evening administration.
  • a morning or evening injectate is typically administered as a single injection (e.g., injection of a 0.7 ml volume).
  • an injectate can be delivered using more than one (e.g., two) injections, as may be done when the injectate volume is greater than comfortably tolerated as a single injection.
  • “/kg” e.g., 7,500 pmol/kg” means“per kilogram patient body weight.”
  • the present invention provides methods and compositions for the treatment of hyperinsulinemic hypoglycemia by subcutaneous (SC) administration of a therapeutically effective dose of exendin(9-39).
  • a patient to be treated according to the methods described herein is a patient having hyperinsulinemic hypoglycemia (HH).
  • the patient having hyperinsulinemic hypoglycemia has previously had bariatric surgery (e.g., Roux-en-Y Gastric Bypass) and/or a related metabolic procedure.
  • the patient has previously had bariatric surgery (e.g., Roux-en-Y Gastric Bypass) and/or a related metabolic procedure and is at risk for developing hyperinsulinemic hypoglycemia.
  • Patients with hyperinsulinemic hypoglycemia may be identified by any suitable method.
  • hyperinsulinemic hypoglycemia is diagnosed by the presence of Whipple’s triad, which has the following criteria: (1) the occurrence of hypoglycemic symptoms; (2) documented low plasma glucose level at the type of the symptoms; and (3) resolution of the symptoms after plasma glucose is raised.
  • hyperinsulinemic hypoglycemia is defined by the occurrence of capillary glucose ⁇ 50 mg/dL at least once per month by patient report or medical record.
  • hyperinsulinemic hypoglycemia is defined by a plasma glucose concentration of ⁇ 55 mg/dL during an oral glucose tolerance test or meal tolerance test in association with inappropriately elevated plasma insulin ( ⁇ 3 uU/mL) or c-peptide (>0.3 mg/dL) when glucose was ⁇ 55 mg/dL.
  • hyperinsulinemic hypoglycemia is defined by a plasma glucose concentration of ⁇ 60 mg/dL during an oral glucose tolerance test or meal tolerance test in association with inappropriately elevated plasma insulin ( ⁇ 3 uU/mL) or c- peptide (>0.3 mg/dL) when glucose was ⁇ 60 mg/dL.
  • Hyperinsulinemic hypoglycemia encompasses the conditions dumping syndrome, nesideoblastosis, noninsulinoma pancreatogenous hypoglycemia syndrome (NIPHS), and/or post-prandial reactive hypoglycemia.
  • Hyperinsulinemic hypoglycemia may result from a gastric or bariatric procedure, such as a Roux-en-Y gastric bypass (RYGB), or may have a congenital, acquired, or induced origin.
  • the patient treated has previously had a bariatric procedure and/or related metabolic procedure, such as a Roux-en-Y Gastric Bypass procedure.
  • Bariatric and/or related metabolic procedures include, but are not limited to, Roux-en-Y Gastric Bypass, Vertical Sleeve Gastrectomy, placement of an endosleeve device, such as the EndoBarrier Gastrointestinal Liner System, also called an“endoluminal liner,” duodenal mucosal resurfacing, also referred to as duodenal ablation, partial bypass of the duodenum, involving duodeno-ileal or duodeno-jejunal anastomosis, vagal nerve blockade, and/or pyloroplasty).
  • Roux-en-Y Gastric Bypass such as the EndoBarrier Gastrointestinal Liner System, also called an“endoluminal liner”
  • duodenal mucosal resurfacing also referred to as duodenal ablation
  • a bariatric procedure typically involves any of the foregoing: partially or completely bypassing the duodenum and/or decreasing nutrient exposure to the duodenum, increasing the rapidity of nutrient transit to the lower part of the intestines (often specifically the ileum), and/or otherwise increasing ileal nutrient exposure.
  • Bariatric surgery may be intended for weight loss or metabolic benefit (such as resolution of diabetes), or both.
  • Such weight loss or metabolic procedures referred to herein as“bariatric procedures” may enhance secretion of GLP-1 from the distal small intestine, especially the ileum, leading to elevated insulin secretion, and in some patients hypoglycemia.
  • the patient may be referred to as a“post bariatric surgery” patient or“post-RYGB.”
  • the patient has previously had a related metabolic procedure.
  • the patient treated has previously had a non-bariatric surgical procedure involving the gastrointestinal system (including but not limited to esophagectomy, for example for treatment of esophageal cancer, Nissen Fundoplication, for example for treatment of gastroesophageal reflux, or gastrectomy, for example for treatment of gastric cancer) and so may be referred to herein as“post gastrointestinal surgery.”
  • the patient treated is prediabetic and/or insulin resistant and may benefit from prevention of pancreatic hyperstimulation from oral carbohydrate ingestion leading to post-prandial hypoglycemia.
  • a treated patient has a congenital, acquired, or induced form of hyperinsulinemic hypoglycemia, such as congenital hyperinsulinism or sometimes referred to as congenital nesidioblastosis.
  • the patient has had bariatric surgery to aid in weight loss and/or metabolic control and has suffered hypoglycemic excursions requiring urgent medical attention; such patients, as demonstrated conclusively in the examples below, can benefit markedly from treatment with a subcutaneously administered formulation of exendin(9-39) in accordance with the invention.
  • a typical adult patient with hyperinsulinemic hypoglycemia will present within 10 years of bariatric and/or other gastrointestinal surgery with symptoms of hypoglycemia (e.g. palpitations, tremor, weakness, sweating, confusion, fatigue, and/or blurred vision) within 5 hours of eating that are associated with a plasma glucose of ⁇ 60 mg/dL and immediate resolution with carbohydrate intake.
  • symptoms of hypoglycemia e.g. palpitations, tremor, weakness, sweating, confusion, fatigue, and/or blurred vision
  • Many patients experience neuroglycopenic symptoms, such as altered mental status, loss of consciousness, or seizures.
  • Hyperinsulinemia (>2 uU/mL or 13.9 pmol/L) may be documented in the proper laboratory setting at the time of the hypoglycemic event. However, documentation of hyperinsulinemia is not always possible due to logistical challenges associated with this testing (which involves induced hypoglycemia) and concerns over patient safety.
  • the methods of the invention provide effective treatment, such that a physician following the same prescribing information herein can expect therapeutic benefit will be achieved in patients whom, for treatment of varying underlying conditions, have had surgical manipulation of the gastrointestinal anatomy, and resultant secondary hyperinsulinemic hypoglycemia.
  • the methods of the invention can be used to treat patients such as: 1) a patient whom, due to gastroesophageal reflux, underwent a Nissen Fundoplication procedure, and subsequently developed secondary hyperinsulinemic hypoglycemia; 2) a patient whom, due to a malignant gastric tumor (e.g.
  • Bilroth I Bilroth II, RYGB, or Jejunal interposition, developed secondary hyperinsulinemic hypoglycemia
  • EGJ esophageal gastric junction
  • endogenous hyperinsulinemia refers to any such condition not caused by bariatric surgery or GI surgery
  • hypoglycemia in these instances can be severe, even life-threatening.
  • Acquired hyperinsulinism may result from insulinomas, autoimmune syndromes, reactive hypoglycemia, adult nesidioblastosis, or gastric dumping syndrome (not due to bariatric or GI surgery).
  • Congenital hyperinsulinism may manifest in the newborn period, or many years later. Accordingly, the methods and formulations of the invention include methods to treat such conditions.
  • a sustained release formulation and/or an immediate release formulation that is administered continuously, such as via a subcutaneous pump, would be employed, with particular emphasis on the prevention of nocturnal hyperinsulinemia.
  • hyperinsulinism may further be induced as a medicinal side-effect of, for example, a GLP-1 agonist, such as exenatide, liraglutide, lixisenatide, albiglutide, and dulaglutide.
  • a GLP-1 agonist such as exenatide, liraglutide, lixisenatide, albiglutide, and dulaglutide.
  • the methods and formulations of the invention include methods to treat overdoses with such drugs.
  • patients with hyperinsulinemic hypoglycemia may also present with cumulative hyperinsulinemic hypoglycemia-associated cognitive impairment.
  • the methods and formulations of the invention include methods to treat or prevent a worsening of cognitive impairment in such patients.
  • acute and chronic hypoglycemia may be associated with morbidities not only such as cognitive impairment, but also depression, heart palpitations/tachycardia, and potentially other conditions, all of which may be reduced or prevented by preventing hypoglycemia by administration of a GLP1A, such as exendin(9-39), as described herein for post-bariatric patients suffering from hyperinsulinemia/hypoglycemia.
  • compositions comprising a therapeutically effective dose of the GLP1A, exendin(9-39), are administered to a patient in need thereof for the treatment or prevention of hyperinsulinemic hypoglycemia.
  • exendin(9-39) is administered by subcutaneous administration (e.g., subcutaneous injection).
  • Sites of injection include, but are not limited to, injection in the thigh, abdomen, upper arm region, or upper buttock region.
  • patients with hyperinsulinemic hypoglycemia may be treated by BID SC administration of exendin(9-39) at therapeutically effective doses of 30 mg or lower (e.g., about 10 mg to 30 mg, 10 mg to 25 mg, 10 mg to 20 mg, 15 mg to 20 mg, 10 mg to 17.5 mg, and 10 mg to 15 mg).
  • Exemplary doses include 10 mg, 12.5 mg, 15 mg, 17.5 mg, 20 mg, 22.5 mg, 25 mg, 27.5 mg or 30 mg.
  • the therapeutically effective amount of exendin(9-39) that is administered is selected from 10 mg, 15 mg, 17.5 mg, and 20 mg.
  • a therapeutically effective dose of exendin(9-39) or range of doses will vary depending upon the needs and physical attributes of the patient. It will be understood by a person of ordinary skill in the art that the doses described herein can be administered at varying concentrations, including but not limited to the injectate concentrations described in Section 3.2.4.1 below.
  • exendin(9-39) may be subcutaneously administered BID to treat hyperinsulinemic hypoglycemia.
  • exendin(9-39) is subcutaneously administered QD.
  • BID (twice per day) administration is well known in the medical arts.
  • BID doses are administered (e.g., self-administered) at about 12 hour intervals (e.g., 7 a.m. and 7 p.m.).
  • shorter (e.g., 8 a.m. and 6 p.m.) or longer (e.g., 7 a.m. and 10 p.m.) intervals between administrations are possible provided the administrations are at least about 6 hours apart.
  • the administrations are at least about 7 hours, 8 hours, 9 hours, 10 hours or 11 hours apart.
  • the administrations are not more than about 15 hours apart.
  • an immediate-release formulation of exendin(9- 39) is provided as a subcutaneous injectable formulation that is administered prior to the administration of a meal.
  • exendin(9-39) is administered within 60-150 minutes (e.g., within 90-120 minutes) prior to morning and evening meals (or before the two main meals of the day, approximately 6 hours or more apart).
  • exendin(9-39) is administered at least one hour prior to the morning meal.
  • the BID dosing will be a morning and evening administration with a morning administration after wakening in the morning and evening administration about 12 hours later (in some embodiments, about 12-14 hours, about 12-16 hours later, or about 9-15 hours later).
  • the morning administration may be before or after the morning meal (breakfast).
  • the dosing schedule is independent of (i.e., not based on, or dictated by) the timing of meals.
  • the morning administration is within a specified time before and/or after the morning meal (e.g. one hour before and/or one hour after breakfast).
  • the morning administration is before or after the morning meal, as discussed above, and the evening administration is prior to retiring for the night (bedtime) such as between the evening meal and bedtime, or within 1, 2, or 3 hours of bedtime.
  • the dosing schedule is semi-independent of mealtimes.
  • the morning dose is administered on a predetermined schedule relative to the morning meal and the evening dose is scheduled at a time independent of the time of the evening meal (e.g., about 12 hours after the morning administration without regard to the time of the evening meal).
  • the schedule, dose, route and formulations of the invention allow the evening administration to provide additional protection at breakfast, and the morning administration to provide protection during the day (e.g., lunch, dinner, or multiple small meals during the day).
  • subcutaneous BID administration of a therapeutically effective dose of exendin(9-39) is protective even when not timed to coincide with meals.
  • an IV bolus injection of 7,500 pmol/kg exendin(9-39) reversed hypoglycemia only if timed to coincide with the peak GLP-1 plasma concentrations.
  • the dosing approaches set forth herein provides considerably more flexibility to the patient than alternative approaches, resulting in increased compliance and a superior quality of life for the patient.
  • certain pharmacokinetic parameters of SC BID exendin(9-39) administration can be modified by selecting the concentration of exendin(9-39) in the injectate.
  • concentration of exendin(9-39) in the injectate As described in the Examples, subcutaneous injection of a low concentration formulation results in a shorter Tmax (i.e., a faster rise to Cmax) relative to a higher concentration.
  • Subcutaneous injection of a high concentration formulation results in a lower Cmax, a longer Tmax, and longer half-life relative to a lower concentration. See Fig.7A and Table 4.
  • a concentration less than 20 mg/ml is a low concentration, e.g., 4-20 mg/ml, preferably about 10-20 mg/ml, and often about 8-16 mg/ml, most often about 13-16 mg/ml, and very often 15 mg/ml.
  • the low concentration formulation results in a pharmacokinetic profile useful for BID administration.
  • a concentration greater than about 20 mg/ml is considered a“high” concentration.
  • a relatively more concentrated solution for example in a range inclusive of and exceeding 20 mg/ml, e.g., 20-40 mg/ml, will result in a lower Cmax, with a longer half-life.
  • subcutaneous administration of exendin(9-39) at a dose of about 20 mg and concentration of about 24 mg/ml exhibited a significantly longer half-life than subcutaneously administered exendin(9- 39) at a dose of about 10 mg and concentration of about 16 mg/ml (9.14 hours vs. 3.60 hours).
  • a more highly concentrated solution of exendin(9-39) results in an exendin(9-39) plasma Cmax that is lower than a relatively lower concentration formulation but which is still greater than a preferred steady state plasma exendin(9-39) concentration of 70 ng/ml or greater (e.g., as shown in Figure 7A and Table 4 for the“10X” (approximately 20 mg) dose as compared to the“5X” (approximately 10 mg) dose).
  • a more concentrated solution may be more amenable to less frequent dosing, e.g., QD dosing, or to BID dosing that is not tied to meals.
  • exendin(9-39) is subcutaneously administered at a concentration of about 4-25 mg/ml, about 4-20 mg/ml, about 10-25 mg/ml, about 10-20 mg/ml, about 10-18 mg/ml, about 8-16 mg/ml, about 12-20 mg/ml, about 10-15 mg/ml, or about 13-16 mg/ml (e.g., about 4 mg/ml, about 5 mg/ml, about 6 mg/ml, about 7 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 11 mg/ml, about 12 mg/ml, about 13 mg/ml, about 14 mg/ml, about 15 mg/ml, about 16 mg/ml, about 17 mg/ml, about 18 mg/ml, about 19 mg/ml, about 20 mg/ml, about 21 mg/ml, about 22 mg/ml, about 23 mg/ml, about 24 mg/ml, or about 25 mg/ml (e.g.
  • exendin(9-39) is subcutaneously administered at a concentration in the range of about 13 mg/ml to about 16 mg/ml. In some embodiments, exendin(9-39) is subcutaneously administered at a concentration of about 15 mg/ml.
  • both a relatively lower dose of 10 mg and a relatively higher dose of 30 mg yielded a Cmax greater than the preferred steady state plasma exendin(9-39) concentration of 70 ng/ml or greater and were efficacious in reversing hyperinsulinemic hypoglycemia when administered at approximately equal concentrations in the range of about 13-16 mg/ml.
  • a relatively lower dose of exendin(9-39) e.g., a dose of about 5-10 mg, e.g., about 5 mg, about 7.5 mg, or about 10 mg
  • a relatively lower dose of exendin(9-39) can be efficacious in treating hyperinsulinemic hypoglycemia by adjusting the exendin(9-39) solution to an appropriate concentration as described herein.
  • a relatively lower dose of exendin(9-39) (e.g., a dose of about 5-10 mg, e.g., about 5 mg, about 7.5 mg, or about 10 mg) is administered at a concentration of at least about 10 mg/ml, e.g., at a concentration in the range of about 13-16 mg/ml, e.g., at a concentration of about 15 mg/ml.
  • exendin(9-39) is subcutaneously administered at a concentration sufficient to result in a steady state plasma exendin(9-39) concentration of at least 70 ng/ml, at least 100 ng/ml, or at least 150 ng/ml as measured by liquid chromatograph-mass spectrometry.
  • exendin(9-39) is subcutaneously administered at a concentration sufficient to result in a steady state plasma exendin(9-39) concentration of about 100-200 ng/ml.
  • exendin(9-39) is subcutaneously administered at a concentration sufficient to result in a steady state plasma exendin(9-39) concentration of at least 70 ng/ml up to 250 ng/ml.
  • each administration of a BID subcutaneous administration of exendin(9-39) results in an exendin(9-39) Cmax of at least 100 ng/ml.
  • each administration of a BID subcutaneous administration of exendin(9-39) results in an exendin(9-39) Cmax of at least 150 ng/ml.
  • the exendin(9-39) injectate comprises an exendin (9- 39) dose and concentration that, when administered, results in steady state plasma exendin(9- 39) concentration of at least 70 ng/ml, preferably at least 100 ng/ml, or even more preferably at least 150 ng/ml, as measured by LCMS.
  • the exendin(9-39) formulation has such a dose and concentration that results in steady state plasma exendin(9- 39) concentration of 100-250 ng/ml, e.g., 100-200 ng/ml, 100-150 ng/ml, or 150-200 ng/ml.
  • each dose is administered in a total volume ranging from 0.25-2 ml injectate, with most patients administering an injection volume ranging from 0.5-1.5 ml, e.g., 0.7-1 ml.
  • Exendin(9-39) may be administered in any pharmaceutically acceptable form.
  • exendin(9-39) is formulated with a pharmaceutically acceptable diluent or carrier that is suitable for subcutaneous administration.
  • pharmaceutically acceptable diluents or carriers include, but are not limited to, water, saline, and isotonic buffer solutions.
  • the injectate formulation further comprises one or more additional excipients such as preservatives and pH adjustment agents.
  • exendin(9-39) is formulated in normal saline (0.9% saline).
  • the exendin(9-39) is formulated with an antimicrobial preservative, a tonicity- adjusting agent, such as mannitol, and/or a buffer (e.g., to bring the solution to a pH of about 4-5).
  • Example 3 and Figure 7A administration of a lower dose (10 mg, or“5x”) resulted in a higher exendin(9-39) Cmax than higher doses formulated in the same volume of solution (i.e., having a higher concentration). Without intending to be bound by a particular mechanism, this may be a result of aggregation (e.g., dimer or higher multimer formation).
  • exendin(9-39) is formulated with an agent to reduce aggregation or dimer formation such as a surfactant (e.g., a non-ionic surfactant, such as a polysorbate or a poloxamer), polyol, or sugar, or by optimizing the pH and/or ionic strength of the solution.
  • a surfactant e.g., a non-ionic surfactant, such as a polysorbate or a poloxamer
  • polyol such as a polysorbate or a poloxamer
  • sugar or by optimizing the pH and/or ionic strength of the solution.
  • exendin(9-39) is formulated for immediate release.
  • exendin(9-39) is formulated as an injectable, immediate-release formulation of exendin(9-39) using a formulation that is used to deliver exenatide, marketed as BYETTATM (see United States Patent Nos.: 5,424,286; 6,858,576; 6,872,700; 6,902,744; 6,956,026; 7,297,761; 7,521,423; and 7,741,269, incorporated herein by reference).
  • exendin(9-39) is formulated for extended release, i.e., an extended release formulation, such that, when administered, the formulation ensures that the active drug product has a lasting presence in the blood throughout the targeted time period in the course of treatment.
  • extended release formulation i.e., an extended release formulation
  • Use of these formulations and methods allows plasma glucose homeostasis to be maintained with fewer subcutaneous injections, relative to immediate release formulations.
  • exendin(9-39) is formulated with microspheres or nano-lipocapsules, which provide for sustained and extended release profiles.
  • exendin(9-39) is formulated with slowly eroding microspheres.
  • microspheres include, for example and without limitation, those made with a biopolymer, such as Poly (lactic-co-glycolic acid) (PLGA) or its equivalent.
  • PLGA Poly (lactic-co-glycolic acid)
  • Such formulations provide for release of drug over an extended period of time (1-10 weeks).
  • exendin(9-39) is loaded into the microspheres, and the formulation provides that exendin is steadily released over time as the matrix materials degrade.
  • These microspheres can be formulated to minimize drug bursts and maintain a steady release profile.
  • exendin(9-39) is encapsulated into nano-lipocapsules to prepare another formulation of the invention, which provides similar sustained and extended drug release.
  • These formulations are provided in a variety of particle and capsule sizes and compositions, providing the physician a variety of rapid, medium, and slow release profile formulations to optimize therapy for individual patients.
  • exendin(9-39) is formulated as an injectable, extended- release formulation of exendin(9-39) using a formulation that is used to deliver exenatide, marketed as BYDUREONTM (see United States Patent Nos.: 5,424,286; 6,479,065; 6,495,164; 6,667,061; 6,824,822; 6,858,576; 6,872,700; 6,956,026; 7,223,440; 7,456,254; 7,563,871; 7,612,176; 7,741,269; 8,216,180; 8,329,648; 8,431,685; 8,439,864; 8,461,105; and 8,906,851, incorporated herein by reference).
  • exendin(9-39) is formulated using a formulation that is used to deliver liraglutide, delivered in a daily dose, marketed as Victoza TM (see U.S. Patent Nos.6,004,297; 6,268,343; 6,458,924; 7,235,627; 8,114,833; and 8,846,612).
  • exendin(9-39) is formulated as a sterile, preserved isotonic solution in a unit or multi-dose glass vial or ampule for administration with the use of a syringe, similar to the glucagon emergency kit.
  • the exendin(9-39) is provided as an injectable suspension in a single-dose tray containing a vial of exendin(9- 39), a vial connector, a prefilled diluent syringe, and one or more needles.
  • exendin(9-39) is formulated as a sterile, preserved isotonic solution in a glass cartridge pen-injector device.
  • Such compositions for example and without limitation contain 5-30 mg of exendin(9-39), an appropriate volume of an antimicrobial preservative, a tonicity-adjusting agent, such as mannitol, and a buffer to bring the solution to a pH of about 4-5.
  • the formulation of exendin(9-39) is provided as an injectable suspension in a single-dose pen containing exendin(9-39), a diluent, and one or more needles.
  • microneedles coated with or containing the formulation of exendin(9-39) are used.
  • exendin(9-39) is formulated as a sterile, preserved isotonic solution in a glass cartridge pen-injector device.
  • Such compositions for example and without limitation contain 5-30 mg of exendin(9-39), an appropriate volume of an antimicrobial preservative, a tonicity-adjusting agent, such as mannitol, and a buffer to bring the solution to a pH of about 4-5.
  • twice-per-day administration comprises administering a morning injectate and an evening injectate that contain different exendin(9-39) doses and/or different concentrations of exendin(9-39).
  • each of the injectates has an exendin(9- 39) amount and concentration within the ranges described herein.
  • the amount of exendin(9-39) in the evening administration is greater than the amount in the morning injectate and/or the exendin(9-39) concentration in the evening injectate is greater than the concentration of exendin(9-39) in the morning injectate.
  • the two injectates will have different quantities, the same concentration, of.
  • the two injectates will have the same amount of exendin(9-39) but different concentration. In some embodiments both the concentration and amount of exendin(9-39) will be different.
  • the increased amount of exendin(9-39) administered in the evening may provide higher exendin(9-39) levels at the time of the morning meal.
  • the increased concentration of exendin(9-39) is expected to result in a more “flat” plasma concentration profile, including a longer time to Tmax, for a more sustained effect at the time of the morning meal.
  • the amount of exendin(9-39) in the evening injectate is 5 mg to 10 mg greater than the amount in the morning injectate. In some embodiments the amount of exendin(9-39) in the evening injectate is 5 mg greater than the amount in the morning injectate. In some embodiments the amount of exendin(9-39) in the evening injectate is 10 mg greater than the amount in the morning injectate. In some embodiments the amount of exendin(9-39) in the morning injectate is 10 mg, 15 mg, or 20 mg.
  • the concentration of exendin(9-39) in the evening injectate is 5 mg/ml-10 mg/ml greater than the amount in the morning injectate. In some embodiments the concentration of exendin(9-39) in the evening injectate is about 5 mg/ml greater than the amount in the morning injectate. In some embodiments the concentration of exendin(9-39) in the evening injectate is about 10 mg/ml greater than the amount in the morning injectate. In some embodiments the concentration of exendin(9-39) in the morning injectate is 10-16 mg/ml and the concentration of exendin(9-39) in the evening injectate is higher and is in the range of 15-20 mg.
  • the amount of exendin(9-39) in the evening injectate is 5 mg-10 mg greater than the amount in the morning injectate. In some such cases the amount of exendin(9-39) in the morning injectate is 10 mg and the amount in the evening injectate is 15 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 10 mg and the amount in the evening injectate is 20 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 15 mg and the amount in the evening injectate is 20 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 15 mg and the amount in the evening injectate is 25 mg.
  • the amount of exendin(9-39) in the morning injectate is 20 mg and the amount in the evening injectate is 25 mg. In some such cases the amount of exendin(9-39) in the morning injectate is 20 mg and the amount in the evening injectate is 30 mg.
  • the concentration of exendin(9-39) in the morning injectate is not the same as the concentration of exendin(9-39) in the evening injectate.
  • the concentration of exendin(9-39) in morning injectate is 15 mg/ml and the concentration of exendin(9-39) in the evening injectate is 20 mg/ml.
  • the exendin(9-39) amount and concentration of the morning and evening injectates are selected such that the exendin(9-39) Tmax after the evening administration is longer than the Tmax after the morning administration.
  • the evening injectate is prepared or formulated to favor multimerization (e.g., dimerization) or precipitation of the exendin(9-39).
  • Administration of the injectate at bedtime can delay absorption, producing a slower release profile compared to the morning administration, resulting in an advantageous basal morning level of at least 30 ng/mL
  • Methods for preparing compositions comprising multimerized proteins are known. For example, the addition of a basic protein, such as protamine, to the exendin(9-39) preparation can favor formation of multimer peptide configurations.
  • multimerization can be achieved by precipitating the exendin(9-39) out of solution, for example through the addition of salts, such as zinc salts, such that the molar ratio of the salt with respect to exendin(9-39) is greater than 1, so as to reduce the solubility of exendin(9-39) in a neutral solvent.
  • salts such as zinc salts
  • raising the pH (for example to 7.4), in the presence of such salts can be used to favor precipitation of the peptide.
  • the level of aggregation or multimerization in the evening injectate is greater than the level in the morning injectate.
  • the exendin(9-39) is in a less soluble form in the evening injectate compared to the morning injectate.
  • Patients may receive therapy for a predetermined time, an indefinite time, or until an endpoint is reached. Treatment may be continued on a continuous daily or weekly basis for at least two to three months, six months, one year, or longer. In some embodiments, therapy is for at least 30 days, at least 60 days, at least 90 days, at least 120 days, at least 150 days, or at least 180 days. In some embodiments, treatment is continued for at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, or at least one year. In some embodiments, treatment is continued for the rest of the patient’s life or until administration is no longer effective in maintaining normal plasma glucose levels to provide meaningful therapeutic benefit. In some embodiments, adult patients (60-100 kg or more) will receive therapeutic benefit from a single dose of exendin(9-39).
  • Devices such as injectable pen devices and pumps suitable for subcutaneous injections are well known. Such devices may be used to deliver the exendin(9-39) formulations described hereinabove according to the methods described herein.
  • exendin(9-39) is administered using an injectable pen device that may be pre-programmed to deliver a fixed dosage amount.
  • the device is pre-programmed to deliver a fixed dosage ranging from 5-30 mg, e.g., 10-20 mg, or 7.5-15 mg, depending upon the needs and physical attributes of the patient.
  • the exendin(9-39) is formulated as an immediate release preparation, and is packaged, for example, in the form of a single or dual-chamber pen device (e.g., a 1 to 5mL dual chamber pen– either a disposable pen or one that reloads disposable cartridges).
  • the drug product can be supplied as a freeze-dried lyophilized powder, stored in a 1 to 3 mL or larger, e.g., 5 mL, dual-chamber cartridge that is compatible with a disposable pen injector (see, for example, the Ypsomed dual chamber cartridge/pen injector: www.ypsomed.com/yds/products/dual-chamber-pens.html).
  • Dose strengths can be conveniently made available to patients, including for example doses in the range of 5-30 mg of exendin(9-39), to be reconstituted in a volume of 0.25-2.0 ml normal saline per dose, or other pharmaceutically acceptable diluent suitable for subcutaneous administration.
  • the drug product is supplied as individual injectable pen devices that are pre-programmed to deliver a fixed dosage amount, in which the morning dosage amount and the evening dosage amount are different amounts and/or concentrations.
  • a first pen e.g., for morning administration
  • delivers a dose in the range of 5-15 mg e.g., a dose of 5 mg, 7.5 mg, 10 mg, 12.5, or 15 mg
  • a second pen e.g., for evening administration
  • delivers a higher dose in the range of 15-20 mg e.g., a dose of 15 mg, 17.5 mg, or 20 mg.
  • the first pen delivers a dose of 10 mg and the second pen delivers a dose of 15 mg.
  • a first pen delivers a dose of 15 mg and the second pen delivers a dose of 20 mg.
  • kits comprising individual injectable pen devices as described herein.
  • a kit comprises a plurality of individual injectable pen devices (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more pens in a kit).
  • the kit comprises two or more individual injectable pen devices that are pre-programmed to deliver a fixed dosage amount, in which the morning dosage amount and the evening dosage amount are different amounts and/or concentrations.
  • the kit comprises a first pen (e.g., for morning administration) that delivers a dose in the range of 5-15 mg (e.g., a dose of 5 mg, 7.5 mg, 10 mg, 12.5, or 15 mg) and further comprises a second pen (e.g., for evening administration) that delivers a higher dose in the range of 15-20 mg (e.g., a dose of 15 mg, 17.5 mg, or 20 mg).
  • the kit comprises a first pen that delivers a dose of 10 mg and a second pen that delivers a dose of 15 mg.
  • the kit comprises a first pen that delivers a dose of 15 mg and a second pen that delivers a dose of 20 mg.
  • the methods of the invention comprise the use of a subcutaneous pump, and the invention provides such pumps containing exendin(9-39) formulated as described herein for subcutaneous delivery.
  • This methodology is generally very convenient for the patient.
  • Compositions for such methods provided by the invention include solution formulations and freeze dried lyophilized powder for reconstitution. See, e.g., Kumareswaran et al., Discovery Medicine, 2012, 13:159-170, incorporated by reference herein.
  • patients treated with the compositions and methods described herein exhibit an improvement in one or more symptoms of hypoglycemia, including but not limited to neuroglycopenic symptoms, beta-adrenergic symptoms, or plasma glucose levels.
  • treatment in the typical adult or pediatric patient refers to treatment such that the postprandial plasma glucose nadir is maintained above a concentration of approximately 55 mg/dl (3.0 mmol/liter) based upon the Endocrine Society's Clinical Guidelines (Journal of Clinical Endocrinology & Metabolism, 2009, 94(3):709-728), and symptoms of hypoglycemia are reduced.
  • normal plasma glucose concentrations are maintained, with those skilled in the art recognizing that in humans a blood glucose level of 65 mg/dl or greater is preferred.
  • treatment in a patient refers to treatment such that at least a 15% increase in postprandial plasma glucose nadir is achieved relative to baseline (e.g., before the onset of treatment).
  • treatment in a patient refers to treatment such that for a patient having a postprandial plasma glucose nadir ⁇ 50 mg/dl at baseline (e.g., before the onset of treatment), an increase in postprandial plasma glucose nadir to ⁇ 55 mg/dl is achieved relative to baseline.
  • Plasma glucose nadir can be measured, for example, by oral glucose tolerance test (OGTT) or meal tolerance test (MTT) as described herein.
  • OGTT oral glucose tolerance test
  • MTT meal tolerance test
  • treatment in a patient refers to treatment such that a statistically significant decrease in the severity of one or more symptoms of hypoglycemia overall during a OGTT or MTT and/or of neuroglycopenic symptoms elicited during the glucose“fall” period of OGTT or MTT is achieved relative to baseline (e.g., before the onset of treatment).
  • Some physicians may desire to treat with a low or initiating (starting) dose (e.g., 5-7.5 mg), escalate to an increased if the initiating dose does not result in acceptable glycemic control, and maintain the initiating dose if glycemic control is sufficient.
  • a low or initiating (starting) dose e.g., 5-7.5 mg
  • a starting dose of 10 mg exendin(9-39) in a morning dose and 10 mg exendin(9-39) in an evening dose is administered to the subject. If this dose does not result in sufficient coverage in the morning (e.g., does not result in sufficient glycemic control at the time of the morning meal), the evening dose may be increased, e.g., to 15 mg exendin(9-39) as the evening dose. In some embodiments, a starting dose of 15 mg exendin(9-39) in a morning dose and 15 mg exendin(9-39) in an evening dose is administered to the subject. If this dose does not result in sufficient coverage in the morning, the evening dose may be increased, e.g., to 20 mg exendin(9-39) as the evening dose.
  • Exendin(9-39) was acquired as a lyophilized peptide: exendin(9-39) acetate 10 mg/vial from Bachem (Clinalfa, Läufelfingen, Switzerland).
  • lyophilized exendin(9-39) was solubilized with 20 ml 0.9% normal saline (NS) for every 10 mg peptide, then diluted in 100 ml 0.9% NS and 50 ml of 25% human serum albumin, in a PVC-free, DEHP-free 1L infusion bag.
  • the bag was covered with an opaque IV bag cover to aid with blinding.
  • An identical- appearing bag was prepared, constituting the placebo infusate, containing the same volume of infusate (NS only) without the presence of peptide or albumin.
  • an IV bolus of 7,500 pmol/kg exendin(9-39) or placebo was administered over 1 minute, while a continuous IV infusion of exendin(9-39) at a rate of 500 pmol/kg/min (providing an infusion dose of about 0.35 mg/kg) or placebo (0.9% saline) was initiated and run for 210 minutes.
  • an OGTT was initiated, wherein patients were instructed to consume a 75 g glucola drink over 20 minutes.
  • Plasma samples were collected at T-40, T+0, T+30, T+45, T+60, T+90, T+105, T+120, T+150, T+180 and at each timepoint immediately taken to the laboratory for processing.
  • the following assays were then conducted: glucose, insulin, GLP-1, GIP, glucagon, and exendin(9-39). If glucose levels dropped to 50 mg/dL or less, the test was stopped and investigators intervened as needed to normalize glucose.
  • T-40 and concomitant with timed blood draws a graded symptom questionnaire was completed repetitively by patients.
  • This questionnaire was adapted from two validated hypoglycemia assessment tools, by segregating symptoms into three clear factors: autonomic, neuroglycopenic, and malaise, and then by adding a severity gradation scale, such that patients rated the severity of each reported symptoms from 1-5 (1: least severe; 5: most severe).
  • Metabolic responses including plasma GLP-1, GIP, and glucagon responses, were measured as shown in Figure 2 A-C, and Table 2.
  • AUC area under the curve
  • continuous exendin(9-39) infusion substantially improved symptoms of hypoglycemia, as demonstrated by the dramatically reduced total hypoglycemic symptom assessment score.
  • two subscores were included: the“Glucose Fall” score, which encompasses symptoms associated with the fall in glucose to nadir, and the“Glucose Rise” score, which encompasses symptoms associated with the rise in glucose to peak.
  • Plasma was assayed at T-40, T+0, T+30, T+45, T+60, T+90, T+105, T+120, T+150, T+180 and at each timepoint immediately taken to the laboratory for processing. Measurements were taken for glucose, insulin, GLP-1, GIP, glucagon, and exendin(9-39).
  • Bioavailability/PK profile of IV exendin(9-39) was evaluated by Cmax, Tmax, AUC0- ⁇ , AUClast, VZ, CL, and T 1/2 .
  • Exendin(9-39) concentration was measured by radioimmunoassay (RIA) as described in Kielgast et al., Diabetes, 2011, 60:1599-1607.
  • FIG. 4A-C As shown in Figures 4A-C, dosing of the IV bolus of exendin(9-39) at 0 minutes or 20 minutes following administration of glucola did not prevent hypoglycemia, whereas dosing at 50 minutes after administration of glucola did prevent hypoglycemia. See, figure legend.
  • Figures 4A-D demonstrates that peak plasma exendin(9-39) concentrations in the range of 500-600 nMol/L by radioimmunoassay at the time of peak plasma GLP-1 concentrations are required to avoid a glucose nadir below 50 mg/dL.
  • the results shown in Figure 4A-D suggest that in the absence of continuous IV infusion, or in the absence of an IV bolus timed precisely to the peak predicted GLP-1 plasma concentrations, hypoglycemia cannot be averted.
  • Exendin(9-39) plasma levels can be measured using a radioimmunoassay (RIA) generally as described in Kielgast et al., Diabetes, 2011, 60:159-1607. Exendin(9-39) plasma levels can be measured using liquid chromatography–mass spectrometry (LCMS) methodology generally as described in Lasaosa et al., J. Chromatogr B Analyt Technol Biomed Life Sci, 2014, 0:186-191. We refer to both methods in the discussion herein, and both methods are used in the scientific literature. We observed that measurement of plasma exendin(9-39) values using RIA were significantly higher than values determined using LCMS. We believe the LCMS values are more accurate. For definitional purposes, a claimed exendin(9-39) concentration (e.g., Cmax) refers to the absolute quantity of Exendin(9-39) which may be determined by LCMS or another equally quantitative method.
  • a claimed exendin(9-39) concentration e.g.,
  • Figure 5A depicts an average of eight patients’ plasma exendin(9-39) concentrations at various timepoints following a 7,500 pmol/kg IV bolus of exendin(9-39) at T-30 minutes, followed by continuous IV fusion at a rate of 500 pmol/kg/min over 210 minutes as described in Example 1. See graph line with error bars. It has also been reported that in healthy subjects an intravenous infusion of exendin(9-39) at 500 pmol/kg/min fully reverses the glucose lowering effect of GLP-1. See, Edwards et al., Diabetes, 1999, 48:86-93.
  • Figure 5A also shows the projected exendin(9-39) plasma concentration that would be expected from administering a single IV bolus of 7,500 pmol/kg exendin(9-39) at T-30 minutes.
  • the half-life of a single dose of intravenously administered exendin(9-39) is about 33.5 minutes (see, Edwards et al., Diabetes, 1999, 48:86- 93).
  • Example 3 A single dose of subcutaneously injected exendin(9-39) effectively reverses hyperinsulinemic hypoglycemia and associated symptoms
  • the IV bolus consisted of 0.025 mg/kg of lyophilized exendin(9-39) (which equates to a dose of approximately 2 mg for an 80 kg patient) solubilized in 20 ml per 10 mg exendin(9-39) (approximately 4 ml normal saline) and then diluted in 100 ml 0.9% normal saline for every 10 mg exendin(9-39) (approximately 20 ml 0.9% normal saline), to which approximately 10 ml 25% human serum albumin was added (50 ml 25% human serum albumin for every 10 mg exendin(9-39)), for a total IV bolus infusion volume of approximately 34 ml.
  • the IV bolus infusion was administered over 1 minute.
  • the subcutaneous injection consisted of 0.025 mg/kg of lyophilized exendin(9-39) (which equates to a dose of approximately 2 mg for an 80 kg patient) solubilized in 0.2 ml normal saline and further diluted in 0.5 ml normal saline to a total volume of 0.7 ml for subcutaneous injection in the arm.
  • Plasma exendin(9-39) concentrations were measured by liquid chromatography–mass spectrometry (LCMS) as described in Lasaosa et al., supra Example 1.
  • SAD single ascending dose
  • Lyophilized exendin(9-39) acetate 10 mg/vial from Bachem (Clinalfa, Läufelfingen, Switzerland) was acquired for each experiment, with each 10 mg vial solubilized in 200 ⁇ l normal saline, then further diluted with normal saline to a total dose of 7,500 pmol/kg, 37,500 pmol/kg, 75,000 pmol/kg, or 112,500 pmol/kg (2.0 mg, 10 mg, 20 mg, or 30 mg of exendin(9-39), respectively, based on a patient weight of 80 kg). The total volume of each injectate was held constant, with further dilution of injectate as required to result in a total volume of injectate of 0.7 ml.
  • the nine subjects were randomized to receive one subcutaneous injection of 7,500 pmol/kg, 37,500 pmol/kg, 75,000 pmol/kg, or 112,500 pmol/kg (2, 10, 20, and 30 mg, respectively, based on an 80 kg patient) in a volume of 0.7 ml normal saline, and four subjects received two or more 0.7 ml injections of 75,000 pmol/kg, or 112,500 pmol/kg in order to maintain an injectate concentration of about 15 mg/ml or less.
  • Plasma samples were collected at T-10, T-0, T+15, T+30, T+45, T+60, T+75, T+90, T+105, T+120, T+135, T+150, T+ 165, T+180, T+210, T+240, T+300, T+480, and T+1440, and at each timepoint the samples were immediately taken to the laboratory for processing.
  • exendin(9-39) administration was efficacious in preventing hypoglycemia, for example as shown by the plasma glucose nadir.
  • Table 5 shows that there was an increasingly favorable PK response with increasing dose, as defined
  • Example 4 Multi-Ascending Dose Trial to assess the efficacy, tolerability, and pharmacokinetic profile of BID exendin(9-39) in patients with post-bariatric hyperinsulinemic hypoglycemia
  • This example describes a Phase 2a clinical study protocol for evaluating the safety, tolerability, efficacy, and pharmacokinetic profile of BID exendin(9-39) administered subcutaneously over 3 days to patients with post-bariatric hyperinsulinemic hypoglycemia.
  • the study is a single-blinded, dose-randomized, cross-over design study that is being conducted at the Stanford University School of Medicine. All subject visits will take place in the Clinical and Translational Research Unit (CTRU). Sixteen to twenty eligible subjects will be assigned to one of five dose levels (2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg) to receive subcutaneous injection of BID exendin(9-39) administered for three days. After a baseline Oral Glucose Tolerance Test (OGTT) is conducted on Day 0 wherein metabolic and symptomatic analyses will occur, subjects will return to the research clinic on Day 1 to initiate a BID dosing schedule for 3 days.
  • CTT Oral Glucose Tolerance Test
  • Randomization/blinding For the first four subjects dosed, the subjects were randomly assigned to one of the following dose levels: 2.5 mg, 5 mg, or 10 mg. The remaining subjects will be randomly assigned to one of the following dose levels: 10 mg, 15 mg, or 20 mg. All subjects will remain blinded throughout. With the exception of the PI and sub-investigator who will remain un-blinded for safety purposes, all site personnel including nurses and study coordinators, who conduct patient symptom surveys, will remain masked to treatment assignment.
  • Oral Glucose Tolerance Test (OGTT): The OGTT will consist of administration of one 75 mg gram glucola drink with 1 gram of crushed acetaminophen to be consumed over 20 minutes.
  • Metabolic glucose, c-peptide, insulin, GLP-1, GIP, glucagon
  • PK AUC 0-720 , C max , T max , T 1/2 , C trough .
  • Anticipated PK profile It was anticipated, based on the prior results for a single subcutaneous injection (as shown in Example 3), that after administration of a 5 mg, 10 mg, 15 mg, or 20 mg dose the plasma concentration of exendin(9-39) would return to ⁇ 20 ng/mL or even close to 0 ng/mL within 720 minutes of injection. However, based on the intermediate results of BID dosing for 3 days as shown in Figure 10 and as discussed below, it is expected that administration of a 10-30 mg dose will result in a higher nadir, such as a nadir of about 30-80 ng/ml within 720 minutes after injection.
  • A“therapeutic benefit” may be defined with reference to effect on plasma glucose.
  • a dosage of exendin(9-39) provides a therapeutic benefit for a patient when the patient has no plasma glucose ⁇ 50 mg/dL at any timepoint from 0-180 minutes during OGTT on Day 3 of treatment as compared to Day 0.
  • a dosage of exendin(9-39) provides a therapeutic benefit for a patient when the patient has at least a 15% increase in plasma glucose nadir during OGTT on Day 3 relative to Day 0.
  • a dosage of exendin(9-39) provides a therapeutic benefit for a patient when the patient has at least a 15% increase in AUC glucose. In some instances a dosage of exendin(9-39) provides a therapeutic benefit for a patient when the patient has a statistically significant decrease in the severity of one or more symptoms of hypoglycemia overall during the OGTT and/or of neuroglycopenic symptoms elicited during the glucose“Fall” period of the OGTT relative to Day 0.
  • a dosage of exendin(9-39) provides a therapeutic benefit for a patient having a plasma glucose nadir ⁇ 50 mg/dL at baseline when the patient exhibits a plasma glucose nadir ⁇ 55 mg/dL after a defined treatment period (e.g., after a 3 day treatment period).
  • Patient 1 was administered a dose of 5 mg at a concentration of 10 mg in 1 ml, subcutaneously administered in the abdomen.
  • a 13.1% increase in AUC glucose was observed as compared to baseline, but hypoglycemia was not prevented, as defined by plasma glucose ⁇ 50 mg/dL.
  • Patient 2 was administered a dose of 2.5 mg at a concentration of 10 mg in 1 ml, subcutaneously administered in the abdomen.
  • an 8.8% increase in AUC glucose was observed, but hypoglycemia was not prevented, as defined by plasma glucose ⁇ 50 mg/dL.
  • Patient 3 was administered a dose of 5 mg at a concentration of 10 mg in 1 ml, subcutaneously administered in the arm.
  • a 16.3% increase in AUC glucose was observed, but hypoglycemia was not prevented, as defined by plasma glucose ⁇ 50 mg/dL.
  • Patient 4 was administered a dose of 10 mg at a concentration of 10 mg in 1 ml, subcutaneously administered in the arm. For this patient, hypoglycemia was not prevented, as defined by plasma glucose ⁇ 50 mg/dL.
  • These intermediate pharmacodynamic results demonstrate an increasing therapeutic benefit, as defined by % increase in glucose AUC with increasing doses administered, with one of the two patients dosed with 5 mg experiencing a greater than 15% increase in AUC glucose as compared to AUC glucose during a baseline oral glucose tolerance test. While hypoglycemia as defined by plasma glucose ⁇ 50 mg/dL was not prevented, a therapeutic dose response was achieved, illustrating that doses of 10-30 mg will result in improved glycemic control, as further shown by Example 3 and Figure 8.
  • a Cmax value is expected to be in the therapeutically effective range of approximately 150-200 ng/ml.
  • Interim pharmacokinetic results from this 3-day trial demonstrate that on average, AUC plasma concentrations increase with increasing days of BID dosing. A higher trough was observed at Day 3 than at Day 1, suggesting several days (e.g., 3-5 days) may be required to reach steady state.
  • the results of this study support efficacy of the 15 mg dose at Day 3 of treatment.
  • the results of this study also support efficacy of the 10 mg dose in less severely disabled patients and/or with longer (e.g., 5 days) treatment.
  • This example demonstrates the method of the invention in which a multi-site multi-ascending dose (MAD) format is used to evaluate the efficacy, safety, and pharmacokinetics of a 28-day trial of immediate release subcutaneous exendin(9-39) administered BID in patients with severe post-bariatric hypoglycemia.
  • the primary objective of this trial is to demonstrate the efficacy of exendin(9-39) on plasma glucose levels during a 3-hour oral Glucose Tolerance Test (OGTT) at the end of 4-week treatment.
  • OGTT oral Glucose Tolerance Test
  • This trial is also intended to demonstrate the efficacy of exendin(9-39) on the frequency and severity of hypoglycemia incidence and associated symptoms in patients with severe post-bariatric hypoglycemia.
  • This trial also demonstrates the pharmacokinetics and pharmacodynamics of exendin(9-39) at each dose level. Furthermore, this trial demonstrates the safety and tolerability profile of the immediate release subcutaneous formulation of exendin(9-39) in patients with severe post-bariatric hypoglycemia.
  • Screening phase All potential subjects will complete an oral glucose tolerance test (OGTT), wherein if plasma glucose falls to less than or equal to 60 mg/dL and all other eligibility criteria are met, the patient will be allowed to enroll in the study. In cases of out of range laboratory values, with the exception of laboratory tests related to re-feeding syndrome, subjects are permitted to re-screen one time.
  • OGTT oral glucose tolerance test
  • RT 4-week randomized treatment
  • All enrolled subjects will participate in a 4-week randomized treatment period wherein subjects will be randomized to one of two exendin(9-39) doses (e.g., 10 mg and 20 mg, 10 mg and 15 mg, or 15 mg and 20 mg) administered BID or matching placebo of the 2 doses.
  • the ratio of treatment assignment to the first exendin(9-39) BID dose, the second exendin(9-39) BID dose, the first matching placebo dose, and the second matching placebo dose will be 2:2:1:1.
  • the subjects will undergo continuous glucose monitoring wearing Dexcoms at home.
  • Open-label extension (OLE) period All patients completing Week 4 of the randomized treatment period and experiencing benefit with exendin(9-39) at the end of RT will be eligible to enter the OLE period.
  • the dose administered will either be an optimal fixed dose level selected at the end of the randomized treatment period of the study or up-titrated to 20 mg BID until any of the following occur: completed 12 months of the open-label extension; unacceptable toxicity; lack of efficacy; protocol deviation; patient withdrew consent; lost to follow-up; death; and study discontinues per the sponsor.
  • the primary efficacy endpoint is measured as the response rate in plasma glucose level at the end of the 4-week RT, defined as the proportion of patients either (1) without plasma glucose ⁇ 55 mg/dL for patients whose glucose nadir is ⁇ 50 mg/dL at baseline OGTT; or (2) without plasma glucose ⁇ 60 mg/dL for patients whose glucose nadir is 55 - ⁇ 60 mg/dL at baseline OGTT.
  • Secondary efficacy endpoints are measured as the improvement in neuroglycopenic symptom score during OGTT at the end of RT (Week 4), where neuroglycopenic symptoms include inability to concentrate, confusion, weakness, drowsiness, dizziness, blurred vision, difficulty speaking (modified from the Edinburgh Hypoglycemia Score, Hepburn 1991); the proportion of patients with severe hypoglycemia during the 4-week RT, where severe hypoglycemia is defined an event requiring assistance of another person to actively administer carbohydrates, glucagon, or take other corrective actions with a blood glucose concentration of ⁇ 50 mg/dL by continuous glucose monitoring (CGM); the proportion of patients with any hypoglycemia event between Week 2 and Week 4, where hypoglycemia is defined as a plasma glucose concentration of ⁇ 55 mg/dL by continuous glucose monitoring (CGM) [Hypoglycemia after Roux-En-Y gastric bypass: detection rates of continuous glucose monitoring (CGM) versus mixed meal test Kefurt 2014]; and the Change
  • the pharmacokinetic and pharmacodynamics endpoints to be measured include C max , T max , T 1/2 , C trough , AUC of exendin(9-39). Exploratory endpoints will include insulin (AUC, Peak, ISR, ICR), GLP-1/GIP, and glucagon concentrations.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Endocrinology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Diabetes (AREA)
  • Emergency Medicine (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Vascular Medicine (AREA)
  • Dermatology (AREA)
  • Dispersion Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)
  • Treatments For Attaching Organic Compounds To Fibrous Goods (AREA)
  • Measurement Of The Respiration, Hearing Ability, Form, And Blood Characteristics Of Living Organisms (AREA)
  • Diaphragms For Electromechanical Transducers (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines Containing Plant Substances (AREA)
PCT/US2016/033837 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with exendin(9-39) WO2016191395A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
AU2016267057A AU2016267057B2 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with exendin(9-39)
ES16800621T ES2887723T3 (es) 2015-05-22 2016-05-23 Tratamiento de hipoglucemia posbariátrica con exendina(9-39)
BR112017025000-4A BR112017025000A2 (pt) 2015-05-22 2016-05-23 tratamento de hipoglicemia pós-bariátrica com exendina (9-39)
LTEPPCT/US2016/033837T LT3297654T (lt) 2015-05-22 2016-05-23 Hipoglikemijos, atsiradusios po bariatrinės chirurgijos, gydymas eksendinu(9-39)
EP21176474.1A EP3936143A1 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with exendin (9-39)
CA3024358A CA3024358A1 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with exendin(9-39)
HRP20211285TT HRP20211285T1 (hr) 2015-05-22 2016-05-23 Liječenje postbarijatrijske hipoglikemije eksendinom (9-39)
SI201631300T SI3297654T1 (sl) 2015-05-22 2016-05-23 Zdravljenje postbariatrične hipoglikemije z eksendinom(9-39)
EP16800621.1A EP3297654B1 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with exendin(9-39)
PL16800621T PL3297654T3 (pl) 2015-05-22 2016-05-23 Leczenie pobariatrycznej hipoglikemii eksendyną (9-39)
RS20211073A RS62368B1 (sr) 2015-05-22 2016-05-23 Lečenje postbarijatrijske hipoglikemije eksendinom(9-39)
DK16800621.1T DK3297654T3 (da) 2015-05-22 2016-05-23 Behandling af post-bariatrisk hypoglykæmi med exendin(9-39)
US15/576,647 US10639354B2 (en) 2015-05-22 2016-05-23 Treatment of hyperinsulinemic hypoglycemia with exendin (9-39)
US16/813,535 US10993991B2 (en) 2015-05-22 2020-03-09 Treatment of post-bariatric hypoglycemia with exendin (9-39)
US17/226,164 US11617782B2 (en) 2015-05-22 2021-04-09 Treatment of post-bariatric hypoglycemia with exendin (9-39)
CY20211100779T CY1124616T1 (el) 2015-05-22 2021-09-01 Θεραπεια της μετα-βαριατρικης υπογλυκαιμιας με εξενδινη(9-39)
AU2022201973A AU2022201973A1 (en) 2015-05-22 2022-03-22 Treatment of post-bariatric hypoglycemia with exendin(9-39)
US18/172,546 US20240000894A1 (en) 2015-05-22 2023-02-22 Treatment of post-bariatric hypoglycemia with exendin (9-39)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201562165743P 2015-05-22 2015-05-22
US62/165,743 2015-05-22
US201562254175P 2015-11-11 2015-11-11
US62/254,175 2015-11-11
US201662329850P 2016-04-29 2016-04-29
US62/329,850 2016-04-29

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/576,647 A-371-Of-International US10639354B2 (en) 2015-05-22 2016-05-23 Treatment of hyperinsulinemic hypoglycemia with exendin (9-39)
US16/813,535 Continuation US10993991B2 (en) 2015-05-22 2020-03-09 Treatment of post-bariatric hypoglycemia with exendin (9-39)

Publications (1)

Publication Number Publication Date
WO2016191395A1 true WO2016191395A1 (en) 2016-12-01

Family

ID=57393681

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2016/033837 WO2016191395A1 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with exendin(9-39)
PCT/US2016/033836 WO2016191394A1 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with glp-1 antagonists

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2016/033836 WO2016191394A1 (en) 2015-05-22 2016-05-23 Treatment of post-bariatric hypoglycemia with glp-1 antagonists

Country Status (17)

Country Link
US (8) US10639354B2 (sl)
EP (4) EP3297654B1 (sl)
AU (4) AU2016267052B2 (sl)
BR (2) BR112017024997A2 (sl)
CA (2) CA3024353A1 (sl)
CL (2) CL2017002911A1 (sl)
CY (2) CY1124616T1 (sl)
DK (2) DK3297653T3 (sl)
ES (2) ES2887723T3 (sl)
HR (2) HRP20211285T1 (sl)
HU (2) HUE055728T2 (sl)
LT (2) LT3297653T (sl)
PL (2) PL3297653T3 (sl)
PT (2) PT3297654T (sl)
RS (2) RS62368B1 (sl)
SI (2) SI3297654T1 (sl)
WO (2) WO2016191395A1 (sl)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018094404A1 (en) 2016-11-21 2018-05-24 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
WO2020081534A1 (en) * 2018-10-15 2020-04-23 Eiger Biopharmaceuticals, Inc. Avexitide for the treatment of hyperinsulinemic hypoglycemia
US10639354B2 (en) 2015-05-22 2020-05-05 The Bot Of The Leland Stanford Junior University Treatment of hyperinsulinemic hypoglycemia with exendin (9-39)
US10653753B2 (en) 2016-03-04 2020-05-19 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
US10987407B2 (en) 2007-01-08 2021-04-27 The Trustees Of The University Of Pennsylvania Methods and compositions for treating hypoglycemic disorders

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019147718A1 (en) * 2018-01-23 2019-08-01 Xeris Pharmaceuticals, Inc. Treatment of post-bariatric hypoglycemia using mini-dose stable glucagon
GB201816639D0 (en) * 2018-10-12 2018-11-28 Heptares Therapeutics Ltd GLP-1 Receptor Antagonist
US20220088144A1 (en) * 2019-01-11 2022-03-24 Universitätsspital Basel Sglt-2 inhibitors or il-1r antagonists for reduction of hypoglycaemia after bariatric surgery
WO2024042518A1 (en) * 2022-08-21 2024-02-29 M. Arkin (1999) Ltd Glp-1 receptor antagonist and methods of use thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US6479065B2 (en) 2000-08-10 2002-11-12 Alkermes Controlled Therapeutics, Inc. Process for the preparation of polymer-based sustained release compositions
US6495164B1 (en) 2000-05-25 2002-12-17 Alkermes Controlled Therapeutics, Inc. I Preparation of injectable suspensions having improved injectability
US6824822B2 (en) 2001-08-31 2004-11-30 Alkermes Controlled Therapeutics Inc. Ii Residual solvent extraction method and microparticles produced thereby
US6858576B1 (en) 1996-08-08 2005-02-22 Amylin Pharmaceuticals, Inc. Methods for regulating gastrointestinal motility
US6872700B1 (en) 1999-01-14 2005-03-29 Amylin Pharmaceuticals, Inc. Methods for glucagon suppression
US6956026B2 (en) 1997-01-07 2005-10-18 Amylin Pharmaceuticals, Inc. Use of exendins for the reduction of food intake
US20080269130A1 (en) * 2007-01-08 2008-10-30 Doris Stoffers Methods and compositions for treating hypoglycemic disorders
US7456254B2 (en) 2004-04-15 2008-11-25 Alkermes, Inc. Polymer-based sustained release device

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SK155694A3 (en) 1992-06-15 1995-05-10 Pfizer Glucagon-like peptide, insulinotropin derivatives, method of their preparation, pharmaceutical agent containing and using these matters
US20040092443A1 (en) 1996-08-06 2004-05-13 Yeda Research And Development Co., Ltd Long-acting exendins and exendin agonists
US6458924B2 (en) 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US7235627B2 (en) 1996-08-30 2007-06-26 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6268343B1 (en) 1996-08-30 2001-07-31 Novo Nordisk A/S Derivatives of GLP-1 analogs
KR100574630B1 (ko) 1998-01-30 2006-04-27 노보 노르디스크 에이/에스 주입 주사기
CA2350887A1 (en) 1998-12-10 2000-06-15 Agouron Pharmaceuticals, Inc. Non-peptide antagonists of glp-1 receptor and methods of use
PT1140145E (pt) 1999-01-14 2005-11-30 Amylin Pharmaceuticals Inc Formulacoes novas de agonistas de exendina e metodos para a sua administracao
US6924264B1 (en) 1999-04-30 2005-08-02 Amylin Pharmaceuticals, Inc. Modified exendins and exendin agonists
US6903074B1 (en) 1999-06-04 2005-06-07 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A.S. Neuromedin b and somatostatin receptor agonists
SE0004462D0 (sv) 2000-12-04 2000-12-04 Pharmacia Ab Novel method and use
WO2002066062A2 (en) 2001-02-01 2002-08-29 Drucker Daniel J Enhancement of glp-2 activity
AU2002257132A1 (en) 2001-04-06 2002-10-21 The Trustees Of The University Of Pennsylvania Erbb interface peptidomimetics and methods of use thereof
CN1635900A (zh) 2001-08-28 2005-07-06 伊莱利利公司 Glp-1和基础胰岛素的预混合物
US7378385B2 (en) 2002-08-08 2008-05-27 University Of Cincinnati Role for GLP-1 to mediate responses to disparate stressors
DK1687019T3 (en) 2003-11-20 2018-02-12 Novo Nordisk As Propylene glycol peptide formulations that are optimal for production and use in injection devices
US8076288B2 (en) 2004-02-11 2011-12-13 Amylin Pharmaceuticals, Inc. Hybrid polypeptides having glucose lowering activity
ES2536235T3 (es) 2004-04-15 2015-05-21 Alkermes Pharma Ireland Limited Dispositivo polimérico de liberación prolongada
DE102004043153B4 (de) 2004-09-03 2013-11-21 Philipps-Universität Marburg Erfindung betreffend GLP-1 und Exendin
JP5198261B2 (ja) 2005-06-06 2013-05-15 カムルス エービー Glp−1類似体製剤
JP5693817B2 (ja) 2005-08-19 2015-04-01 アミリン・ファーマシューティカルズ, リミテッド・ライアビリティ・カンパニーAmylin Pharmaceuticals, Llc 糖尿病の治療法および体重の減少法
EP2636680A3 (en) 2006-05-26 2013-12-11 Amylin Pharmaceuticals, LLC Composition and methods for treatment of congestive heart failure
WO2008122132A1 (de) 2007-04-05 2008-10-16 Tecpharma Licensing Ag Verabreichungsvorrichtung mit funktionalem antriebsglied
DE102007016811A1 (de) 2007-04-05 2008-10-09 Tecpharma Licensing Ag Vorrichtung zur Verabreichung eines fluiden Wirkstoffes aus einer Mehrkammerampulle
CN105688191A (zh) 2007-04-23 2016-06-22 精达制药公司 促胰岛素释放肽的混悬制剂及其应用
CN105903005A (zh) 2008-05-05 2016-08-31 奥拉姆德有限公司 用于口服给予艾塞那肽的方法和组合物
ES2552646T3 (es) 2008-05-21 2015-12-01 Amylin Pharmaceuticals, Inc. Exendinas para disminuir el colesterol y los triglicéridos
EP2303226B1 (en) 2008-06-25 2016-03-23 Endo Pharmaceuticals Solutions Inc. Sustained delivery of exenatide and other polypeptides
EP2310042B1 (en) 2008-07-08 2012-12-05 Novartis AG Use of pasireotide for the treatment of endogenous hyperinsulinemic hypoglycemia
ES2809178T5 (es) 2008-09-04 2024-04-29 Amylin Pharmaceuticals Llc Formulaciones de liberación sostenida que usan portadores no acuosos
PL2413952T3 (pl) 2009-04-03 2017-08-31 Nestec S.A. Polepszenie promowania zdrowego doganiania wzrostu
ES2705249T3 (es) 2009-06-08 2019-03-22 Amunix Operating Inc Polipéptidos reguladores de glucosa y métodos para su producción y uso
WO2011109787A1 (en) 2010-03-05 2011-09-09 Conjuchem, Llc Methods of administering insulinotropic peptides
US20130137645A1 (en) 2010-07-19 2013-05-30 Mary S. Rosendahl Modified peptides and proteins
US8969293B2 (en) 2010-12-23 2015-03-03 Rani Therapeutics, Llc Therapeutic agent preparations comprising exenatide for delivery into a lumen of the intestinal tract using a swallowable drug delivery device
US20120208755A1 (en) 2011-02-16 2012-08-16 Intarcia Therapeutics, Inc. Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers
WO2013059323A1 (en) 2011-10-18 2013-04-25 Prolynx Llc Peg conjugates of exenatide
US20150005233A1 (en) 2012-02-08 2015-01-01 Seneb Biosciences, Inc. Treatment of hypoglycemia
EP3033355A1 (en) 2013-08-16 2016-06-22 Medimmune Limited Gip and glp-1 receptor dual-agonists for the treatment of diabetes
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
EP3297654B1 (en) 2015-05-22 2021-07-07 The Board of Trustees of the Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin(9-39)
WO2017152014A1 (en) 2016-03-04 2017-09-08 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
IL266568B1 (en) 2016-11-21 2024-05-01 Eiger Biopharmaceuticals Inc Conditioned formulations of Exendin (39-9)

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US6858576B1 (en) 1996-08-08 2005-02-22 Amylin Pharmaceuticals, Inc. Methods for regulating gastrointestinal motility
US6956026B2 (en) 1997-01-07 2005-10-18 Amylin Pharmaceuticals, Inc. Use of exendins for the reduction of food intake
US6872700B1 (en) 1999-01-14 2005-03-29 Amylin Pharmaceuticals, Inc. Methods for glucagon suppression
US6495164B1 (en) 2000-05-25 2002-12-17 Alkermes Controlled Therapeutics, Inc. I Preparation of injectable suspensions having improved injectability
US6667061B2 (en) 2000-05-25 2003-12-23 Alkermes Controlled Therapeutics, Inc. Preparation of injectable suspensions having improved injectability
US6479065B2 (en) 2000-08-10 2002-11-12 Alkermes Controlled Therapeutics, Inc. Process for the preparation of polymer-based sustained release compositions
US6824822B2 (en) 2001-08-31 2004-11-30 Alkermes Controlled Therapeutics Inc. Ii Residual solvent extraction method and microparticles produced thereby
US7223440B2 (en) 2001-08-31 2007-05-29 Alkermes, Inc. Residual solvent extraction method and microparticles produced thereby
US7456254B2 (en) 2004-04-15 2008-11-25 Alkermes, Inc. Polymer-based sustained release device
US20080269130A1 (en) * 2007-01-08 2008-10-30 Doris Stoffers Methods and compositions for treating hypoglycemic disorders

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
CHEN ET AL., INTERSPECIES MODELING PHARM RES., vol. 30, 2013, pages 751 - 760
EDWARDS ET AL., DIABETES, vol. 48, 1999, pages 86 - 93
GARDINER ET AL., JPET, vol. 316, 2006, pages 852 - 859
JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM, vol. 94, no. 3, 2009, pages 709 - 728
KIELGAST ET AL., DIABETES, vol. 60, 2011, pages 1599 - 1607
LASAOSA ET AL., J. CHROMATOGR B ANALYT TECHNOL BIOMED LIFE SCI, vol. 0, 2014, pages 186 - 191
MANNING, S ET AL.: "GLP-1: A Mediator of the Beneficial Metabolic Effects of Bariatric Surgery?", PHYSIOLOGY, vol. 30, no. 1, 5 January 2015 (2015-01-05), pages 1 - 28, 50-62, XP055334065, Retrieved from the Internet <URL:http://physiologyonline.physiology.org/content/30/1/50> [retrieved on 20160717] *
MONTROSE-RAFIZADEH ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 272, 1997, pages 21201 - 21206
MYINT ET AL., EUROPEAN JOURNAL OF ENDOCRINOLOGY, vol. 166, 2012, pages 951 - 955
SALEHI ET AL.: "Blockade of Glucagon-like Peptide 1 Receptor Corrects Postprandial Hypoglycemia After Gastric Bypass", GASTROENTEROLOGY, vol. 146, 2014, pages 669 - 680
SALEHI, M ET AL.: "Blockade of Glucagon-like Peptide 1 Receptor Corrects Postprandial Hypoglycemia After Gastric Bypass", GASTROENTEROLOGY, vol. 146, 2014, pages 669 - 680, XP055332926 *
SALEHI, M ET AL.: "Blockade of Glucagon-like Peptide 1 Receptor Corrects Postprandinal Hypoglycemia After Gastric Bypass", GASTROENTEROLOGY, vol. 146, pages 669 - 680,1014

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10987407B2 (en) 2007-01-08 2021-04-27 The Trustees Of The University Of Pennsylvania Methods and compositions for treating hypoglycemic disorders
US10993992B2 (en) 2015-05-22 2021-05-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with GLP-1 antagonists
US11622995B2 (en) 2015-05-22 2023-04-11 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with GLP-1 antagonists
US10639354B2 (en) 2015-05-22 2020-05-05 The Bot Of The Leland Stanford Junior University Treatment of hyperinsulinemic hypoglycemia with exendin (9-39)
US11617782B2 (en) 2015-05-22 2023-04-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin (9-39)
US10660937B2 (en) 2015-05-22 2020-05-26 The Board Of Trustees Of The Leland Stanford Junio Treatment of hyperinsulinemic hypoglycemia with GLP-1 antagonist exendin(9-39)
US10993991B2 (en) 2015-05-22 2021-05-04 The Board Of Trustees Of The Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin (9-39)
US10653753B2 (en) 2016-03-04 2020-05-19 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
US11116820B2 (en) 2016-03-04 2021-09-14 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
US11020484B2 (en) 2016-11-21 2021-06-01 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
WO2018094404A1 (en) 2016-11-21 2018-05-24 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
US11738086B2 (en) 2016-11-21 2023-08-29 Eiger Biopharmaceuticals, Inc. Methods of using buffered formulations of exendin (9-39)
CN112867730A (zh) * 2018-10-15 2021-05-28 艾格尔峰生物制药有限公司 用于治疗高胰岛素低血糖症的阿维西特
WO2020081534A1 (en) * 2018-10-15 2020-04-23 Eiger Biopharmaceuticals, Inc. Avexitide for the treatment of hyperinsulinemic hypoglycemia
EP4233842A3 (en) * 2018-10-15 2023-10-25 Eiger Biopharmaceuticals, Inc. Avexitide for the treatment of hyperinsulinemic hypoglycemia

Also Published As

Publication number Publication date
EP3297653B1 (en) 2021-09-15
US10993992B2 (en) 2021-05-04
BR112017024997A2 (pt) 2018-07-31
US20210290731A1 (en) 2021-09-23
US10660937B2 (en) 2020-05-26
US20240000894A1 (en) 2024-01-04
US11622995B2 (en) 2023-04-11
AU2016267052A1 (en) 2017-12-14
CA3024358A1 (en) 2016-12-01
EP3297653A4 (en) 2018-12-26
SI3297653T1 (sl) 2022-01-31
CL2017002913A1 (es) 2018-06-01
RS62627B1 (sr) 2021-12-31
US20200276272A1 (en) 2020-09-03
EP3297654B1 (en) 2021-07-07
AU2016267057B2 (en) 2021-12-23
LT3297653T (lt) 2022-01-10
AU2016267057A1 (en) 2017-12-14
BR112017025000A2 (pt) 2018-08-07
HRP20211285T1 (hr) 2022-01-07
EP3297654A4 (en) 2018-04-11
HUE055728T2 (hu) 2021-12-28
ES2887723T3 (es) 2021-12-27
CY1124616T1 (el) 2022-07-22
US11617782B2 (en) 2023-04-04
US10993991B2 (en) 2021-05-04
ES2897955T3 (es) 2022-03-03
EP3297653A1 (en) 2018-03-28
US20210315974A1 (en) 2021-10-14
US20180117122A1 (en) 2018-05-03
PT3297653T (pt) 2021-10-25
LT3297654T (lt) 2021-10-25
AU2022201973A1 (en) 2022-04-14
EP3978011A1 (en) 2022-04-06
EP3297654A1 (en) 2018-03-28
US20180147261A1 (en) 2018-05-31
SI3297654T1 (sl) 2021-11-30
US20200276271A1 (en) 2020-09-03
CL2017002911A1 (es) 2018-05-25
DK3297654T3 (da) 2021-09-06
CA3024353A1 (en) 2016-12-01
US10639354B2 (en) 2020-05-05
AU2022202506A1 (en) 2022-05-12
US20230346890A1 (en) 2023-11-02
WO2016191394A1 (en) 2016-12-01
CY1125147T1 (el) 2023-03-24
HUE057301T2 (hu) 2022-05-28
EP3936143A1 (en) 2022-01-12
RS62368B1 (sr) 2021-10-29
PL3297653T3 (pl) 2022-01-31
PT3297654T (pt) 2021-09-06
HRP20211768T1 (hr) 2022-02-18
PL3297654T3 (pl) 2021-12-20
AU2016267052B2 (en) 2022-01-20
DK3297653T3 (da) 2021-10-18

Similar Documents

Publication Publication Date Title
US11617782B2 (en) Treatment of post-bariatric hypoglycemia with exendin (9-39)
US20210315995A1 (en) Buffered Formulations Of Exendin (9-39)
US11116820B2 (en) Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
Kuritzky et al. Safety and efficacy of dulaglutide, a once weekly GLP-1 receptor agonist, for the management of type 2 diabetes

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16800621

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15576647

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016267057

Country of ref document: AU

Date of ref document: 20160523

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017025000

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112017025000

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20171122

ENP Entry into the national phase

Ref document number: 3024358

Country of ref document: CA